Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 8(82): eadd8454, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37083450

ABSTRACT

Our understanding of tissue-resident memory T (TRM) cell biology has been largely developed from acute infection models in which antigen is cleared and sterilizing immunity is achieved. Less is known about TRM cells in the context of chronic antigen persistence and inflammation. We investigated factors that underlie TRM maintenance in a kidney transplantation model in which TRM cells drive rejection. In contrast to acute infection, we found that TRM cells declined markedly in the absence of cognate antigen, antigen presentation, or antigen sensing by the T cells. Depletion of graft-infiltrating dendritic cells or interruption of antigen presentation after TRM cells were established was sufficient to disrupt TRM maintenance and reduce allograft pathology. Likewise, removal of IL-15 transpresentation or of the IL-15 receptor on T cells during TRM maintenance led to a decline in TRM cells, and IL-15 receptor blockade prevented chronic rejection. Therefore, antigen and IL-15 presented by dendritic cells play nonredundant key roles in CD8 TRM cell maintenance in settings of antigen persistence and inflammation. These findings provide insights that could lead to improved treatment of chronic transplant rejection and autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-15 , Humans , Antigens , Inflammation , Memory T Cells
2.
Immunity ; 56(2): 369-385.e6, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36720219

ABSTRACT

In allogeneic hematopoietic stem cell transplantation, donor αß T cells attack recipient tissues, causing graft-versus-host disease (GVHD), a major cause of morbidity and mortality. A central question has been how GVHD is sustained despite T cell exhaustion from chronic antigen stimulation. The current model for GVHD holds that disease is maintained through the continued recruitment of alloreactive effectors from blood into affected tissues. Here, we show, using multiple approaches including parabiosis of mice with GVHD, that GVHD is instead primarily maintained locally within diseased tissues. By tracking 1,203 alloreactive T cell clones, we fitted a mathematical model predicting that within each tissue a small number of progenitor T cells maintain a larger effector pool. Consistent with this, we identified a tissue-resident TCF-1+ subpopulation that preferentially engrafted, expanded, and differentiated into effectors upon adoptive transfer. These results suggest that therapies targeting affected tissues and progenitor T cells within them would be effective.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mice , Animals , T-Lymphocytes , Transplantation, Homologous/adverse effects , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods
3.
J Allergy Clin Immunol ; 150(1): 114-130, 2022 07.
Article in English | MEDLINE | ID: mdl-35085664

ABSTRACT

BACKGROUND: Allergic contact dermatitis (CD) is a chronic inflammatory skin disease caused by type 1 biased adaptive immunity for which there is an unmet need for antigen (Ag)-specific immunotherapies. Exposure to skin sensitizers stimulates secretion of the proinflammatory neuropeptides substance P and hemokinin 1, which signal via the neurokinin-1 receptor (NK1R) to promote the innate and adaptive immune responses of CD. Accordingly, mice lacking the NK1R develop impaired CD. Nonetheless, the role and therapeutic opportunities of targeting the NK1R in CD remain to be elucidated. OBJECTIVE: We sought to develop an Ag-specific immunosuppressive approach to treat CD by skin codelivery of hapten and NK1R antagonists integrated in dissolvable microneedle arrays (MNA). METHODS: In vivo mouse models of contact hypersensitivity and ex vivo models of human skin were used to delineate the effects and mechanisms of NK1R signaling and the immunosuppressive effects of the contact sensitizer NK1R antagonist MNA in CD. RESULTS: We demonstrated in mice that CD requires NK1R signaling by substance P and hemokinin 1. Specific deletion of the NK1R in keratinocytes and dendritic cells, but not in mast cells, prevented CD. Skin codelivery of hapten or Ag MNA inhibited neuropeptide-mediated skin inflammation in mouse and human skin, promoted deletion of Ag-specific effector T cells, and increased regulatory T cells, which prevented CD onset and relapses locally and systemically in an Ag-specific manner. CONCLUSIONS: Immunoregulation by engineering localized skin neuroimmune networks can be used to treat cutaneous diseases that like CD are caused by type 1 immunity.


Subject(s)
Dermatitis, Allergic Contact , Neurokinin-1 Receptor Antagonists , Animals , Dermatitis, Allergic Contact/drug therapy , Haptens , Mice , Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-1 , Substance P
4.
J Heart Lung Transplant ; 40(7): 557-561, 2021 07.
Article in English | MEDLINE | ID: mdl-33958265

ABSTRACT

Successful allogeneic transplantation has been made possible by suppressing activation of the adaptive immune system. Current immunosuppressive therapy prevents rejection by targeting T and B cells. Despite this effective treatment, it is the innate immune system, which includes dendritic cells, monocytes, natural killer cells, that is responsible for the initiation of the adaptive immune response. Recent work has described that the innate immune system is capable of recognizing allogeneic nonself and some of the mechanisms of innate allorecognition have been uncovered. Better understanding of the role of the innate immune system in initiation and maintenance of the allo-immune response has potential to lead to better treatment strategies for transplant patients, prolonging allograft survival. Here, we review advances in our understanding of innate allorecognition in transplantation.


Subject(s)
Graft Rejection/immunology , Immunity, Innate/immunology , Organ Transplantation , Humans , Transplantation, Homologous
5.
Sci Transl Med ; 13(585)2021 03 17.
Article in English | MEDLINE | ID: mdl-33731430

ABSTRACT

Despite the role of donor-specific antibodies (DSAs) in recognizing major histocompatibility complex (MHC) antigens and mediating transplant rejection, how and where recipient B cells in lymphoid tissues encounter donor MHC antigens remains unclear. Contrary to the dogma, we demonstrated here that migration of donor leukocytes out of skin or heart allografts is not necessary for B or T cell allosensitization in mice. We found that mouse skin and cardiac allografts and human skin grafts release cell-free donor MHC antigens via extracellular vesicles (EVs) that are captured by subcapsular sinus (SCS) macrophages in lymph nodes or analog macrophages in the spleen. Donor EVs were transported across the SCS macrophages, and donor MHC molecules on the EVs were recognized by alloreactive B cells. This triggered B cell activation and DSA production, which were both prevented by SCS macrophage depletion. These results reveal an unexpected role for graft-derived EVs and open venues to interfere with EV biogenesis, trafficking, or function to restrain priming or reactivation of alloreactive B cells.


Subject(s)
Extracellular Vesicles , Heart Transplantation , Animals , B-Lymphocytes , Graft Rejection , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
6.
Sci Immunol ; 6(57)2021 03 19.
Article in English | MEDLINE | ID: mdl-33741656

ABSTRACT

Tissue-resident memory T cells (TRM) contained at sites of previous infection provide local protection against reinfection. Whether they form and function in organ transplants where cognate antigen persists is unclear. This is a key question in transplantation as T cells are detected long term in allografts, but it is not known whether they are exhausted or are functional memory T cells. Using a mouse model of kidney transplantation, we showed that antigen-specific and polyclonal effector T cells differentiated in the graft into TRM and subsequently caused allograft rejection. TRM identity was established by surface phenotype, transcriptional profile, and inability to recirculate in parabiosis and retransplantation experiments. Graft TRM proliferated locally, produced interferon-γ upon restimulation, and their in vivo depletion attenuated rejection. The vast majority of antigen-specific and polyclonal TRM lacked phenotypic and transcriptional exhaustion markers. Single-cell analysis of graft T cells early and late after transplantation identified a transcriptional program associated with transition to the tissue-resident state that could serve as a platform for the discovery of therapeutic targets. Thus, recipient effector T cells differentiate into functional graft TRM that maintain rejection locally. Targeting these TRM could improve renal transplant outcomes.


Subject(s)
Allografts/immunology , Antigens/immunology , Graft Rejection/immunology , Immunologic Memory , Memory T Cells/immunology , Allografts/metabolism , Allografts/pathology , Animals , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Graft Rejection/metabolism , Graft Rejection/pathology , Immunohistochemistry , Immunophenotyping , Kidney Transplantation , Memory T Cells/metabolism , Mice , Organ Specificity/genetics , Organ Specificity/immunology , Organ Transplantation/adverse effects , Organ Transplantation/methods , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
7.
J Clin Invest ; 130(10): 5397-5412, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32644975

ABSTRACT

Alarmins, sequestered self-molecules containing damage-associated molecular patterns, are released during tissue injury to drive innate immune cell proinflammatory responses. Whether endogenous negative regulators controlling early immune responses are also released at the site of injury is poorly understood. Herein, we establish that the stromal cell-derived alarmin interleukin 33 (IL-33) is a local factor that directly restricts the proinflammatory capacity of graft-infiltrating macrophages early after transplantation. By assessing heart transplant recipient samples and using a mouse heart transplant model, we establish that IL-33 is upregulated in allografts to limit chronic rejection. Mouse cardiac transplants lacking IL-33 displayed dramatically accelerated vascular occlusion and subsequent fibrosis, which was not due to altered systemic immune responses. Instead, a lack of graft IL-33 caused local augmentation of proinflammatory iNOS+ macrophages that accelerated graft loss. IL-33 facilitated a metabolic program in macrophages associated with reparative and regulatory functions, and local delivery of IL-33 prevented the chronic rejection of IL-33-deficient cardiac transplants. Therefore, IL-33 represents what we believe is a novel regulatory alarmin in transplantation that limits chronic rejection by restraining the local activation of proinflammatory macrophages. The local delivery of IL-33 in extracellular matrix-based materials may be a promising biologic for chronic rejection prophylaxis.


Subject(s)
Graft Rejection/immunology , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Interleukin-33/immunology , Macrophages/immunology , Alarmins/immunology , Allografts , Animals , Child , Disease Models, Animal , Graft Rejection/etiology , Graft Survival/immunology , Humans , Interleukin-33/administration & dosage , Interleukin-33/deficiency , Interleukin-33/genetics , Macrophage Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Myocardium/immunology , Myocardium/pathology , Up-Regulation
8.
Front Immunol ; 11: 918, 2020.
Article in English | MEDLINE | ID: mdl-32547540

ABSTRACT

Over the past few decades, we have witnessed a decline in the rates of acute rejection without significant improvement in chronic rejection. Current treatment strategies principally target the adaptive immune response and not the innate response. Therefore, better understanding of innate immunity in transplantation and how to target it is highly desirable. Here, we review the latest advances in innate immunity in transplantation focusing on the roles and mechanisms of innate allorecognition and memory in myeloid cells. These novel concepts could explain why alloimmune response do not abate over time and shed light on new molecular pathways that can be interrupted to prevent or treat chronic rejection.


Subject(s)
Graft Rejection/immunology , Graft Survival , Immunity, Innate , Immunologic Memory , Isoantigens/immunology , Organ Transplantation/adverse effects , Transplantation Tolerance , Animals , Graft Rejection/prevention & control , Graft Survival/drug effects , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Innate/drug effects , Immunologic Memory/drug effects , Immunosuppressive Agents/therapeutic use , Transplantation Tolerance/drug effects , Treatment Outcome
9.
Science ; 368(6495): 1122-1127, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32381589

ABSTRACT

Immunological memory specific to previously encountered antigens is a cardinal feature of adaptive lymphoid cells. However, it is unknown whether innate myeloid cells retain memory of prior antigenic stimulation and respond to it more vigorously on subsequent encounters. In this work, we show that murine monocytes and macrophages acquire memory specific to major histocompatibility complex I (MHC-I) antigens, and we identify A-type paired immunoglobulin-like receptors (PIR-As) as the MHC-I receptors necessary for the memory response. We demonstrate that deleting PIR-A in the recipient or blocking PIR-A binding to donor MHC-I molecules blocks memory and attenuates kidney and heart allograft rejection. Thus, innate myeloid cells acquire alloantigen-specific memory that can be targeted to improve transplant outcomes.


Subject(s)
Graft Rejection/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Innate , Immunologic Memory , Macrophages/immunology , Monocytes/immunology , Receptors, Immunologic/physiology , Animals , Gene Deletion , Graft Rejection/genetics , Heart Transplantation , Kidney Transplantation , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Receptors, Immunologic/genetics
10.
J Clin Invest ; 130(1): 287-294, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31763998

ABSTRACT

Activation of host T cells that mediate allograft rejection is a 2-step process. The first occurs in secondary lymphoid organs where T cells encounter alloantigens presented by host DCs and differentiate to effectors. Antigen presentation at these sites occurs principally via transfer of intact, donor MHC-peptide complexes from graft cells to host DCs (cross-dressing) or by uptake and processing of donor antigens into allopeptides bound to self-MHC molecules (indirect presentation). The second step takes place in the graft, where effector T cells reengage with host DCs before causing rejection. How host DCs present alloantigens to T cells in the graft is not known. Using mouse islet and kidney transplantation models, imaging cytometry, and 2-photon intravital microscopy, we demonstrate extensive cross-dressing of intragraft host DCs with donor MHC-peptide complexes that occurred early after transplantation, whereas host DCs presenting donor antigen via the indirect pathway were rare. Cross-dressed DCs stably engaged TCR-transgenic effector CD8+ T cells that recognized donor antigen and were sufficient for sustaining acute rejection. In the chronic kidney rejection model, cross-dressing declined over time but was still conspicuous 8 weeks after transplantation. We conclude that cross-dressing of host DCs with donor MHC molecules is a major antigen presentation pathway driving effector T cell responses within allografts.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Graft Rejection/immunology , Islets of Langerhans Transplantation/immunology , Kidney Transplantation , Lymphocyte Activation , Allografts , Animals , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Graft Rejection/pathology , Mice , Mice, Knockout , Transplantation Immunology
11.
J Am Soc Nephrol ; 29(6): 1596-1600, 2018 06.
Article in English | MEDLINE | ID: mdl-29654214

ABSTRACT

Kidney transplantation is the treatment of choice for ESRD but is complicated by the response of the recipient's immune system to nonself histocompatibility antigens on the graft, resulting in rejection. Multiphoton intravital microscopy, referred to as four-dimensional imaging because it records dynamic events in three-dimensional tissue volumes, has emerged as a powerful tool to study immunologic processes in living animals. Here, we will review advances in understanding the complex mechanisms of T cell-mediated rejection made possible by four-dimensional imaging of mouse renal allografts. We will summarize recent data showing that activated (effector) T cell migration to the graft is driven by cognate antigen presented by dendritic cells that surround and penetrate peritubular capillaries, and that T cell-dendritic cell interactions persist in the graft over time, maintaining the immune response in the tissue.


Subject(s)
Graft Rejection/diagnostic imaging , Intravital Microscopy/methods , Microscopy, Fluorescence, Multiphoton , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , Cell Movement , Dendritic Cells/immunology , Graft Rejection/immunology , Mice
12.
Sci Immunol ; 2(12)2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28783664

ABSTRACT

Mice devoid of T, B, and natural killer (NK) cells distinguish between self and allogeneic nonself despite the absence of an adaptive immune system. When challenged with an allograft, they mount an innate response characterized by accumulation of mature, monocyte-derived dendritic cells (DCs) that produce interleukin-12 and present antigen to T cells. However, the molecular mechanisms by which the innate immune system detects allogeneic nonself to generate these DCs are not known. To address this question, we studied the innate response of Rag2-/- γc-/- mice, which lack T, B, and NK cells, to grafts from allogeneic donors. By positional cloning, we identified that donor polymorphism in the gene encoding signal regulatory protein α (SIRPα) is a key modulator of the recipient's innate allorecognition response. Donors that differed from the recipient in one or both Sirpa alleles elicited an innate alloresponse. The response was mediated by binding of donor SIRPα to recipient CD47 and was modulated by the strength of the SIRPα-CD47 interaction. Therefore, sensing SIRPα polymorphism by CD47 provides a molecular mechanism by which the innate immune system distinguishes between self and allogeneic nonself independently of T, B, and NK cells.

14.
Kidney Int ; 90(4): 818-27, 2016 10.
Article in English | MEDLINE | ID: mdl-27528552

ABSTRACT

Bony fish are among the first vertebrates to possess an innate and adaptive immune system. In these species, the kidney has a dual function: filtering solutes similar to mammals and acting as a lymphoid organ responsible for hematopoiesis and antigen processing. Recent studies have shown that the mammalian kidney has an extensive network of mononuclear phagocytes, whose function is not fully understood. Here, we employed two-photon intravital microscopy of fluorescent reporter mice to demonstrate that renal dendritic cells encase the microvasculature in the cortex, extend dendrites into the peritubular capillaries, and sample the blood for antigen. We utilized a mouse model of systemic bacterial infection as well as immune complexes to demonstrate antigen uptake by renal dendritic cells. As a consequence, renal dendritic cells mediated T-cell migration into the kidney in an antigen-dependent manner in the setting of bacterial infection. Thus, renal dendritic cells may be uniquely positioned to play an important role not only in surveillance of systemic infection but also in local infection and autoimmunity.


Subject(s)
Autoimmunity , Bacterial Infections/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Kidney/immunology , T-Lymphocytes/physiology , Animals , Antigen Presentation/immunology , Antigen-Antibody Complex , Dendritic Cells/ultrastructure , Intravital Microscopy , Kidney/blood supply , Kidney/cytology , Kidney/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Models, Animal
15.
Nat Commun ; 7: 12623, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27554168

ABSTRACT

Successful engraftment of organ transplants has traditionally relied on preventing the activation of recipient (host) T cells. Once T-cell activation has occurred, however, stalling the rejection process becomes increasingly difficult, leading to graft failure. Here we demonstrate that graft-infiltrating, recipient (host) dendritic cells (DCs) play a key role in driving the rejection of transplanted organs by activated (effector) T cells. We show that donor DCs that accompany heart or kidney grafts are rapidly replaced by recipient DCs. The DCs originate from non-classical monocytes and form stable, cognate interactions with effector T cells in the graft. Eliminating recipient DCs reduces the proliferation and survival of graft-infiltrating T cells and abrogates ongoing rejection or rejection mediated by transferred effector T cells. Therefore, host DCs that infiltrate transplanted organs sustain the alloimmune response after T-cell activation has already occurred. Targeting these cells provides a means for preventing or treating rejection.


Subject(s)
Dendritic Cells/immunology , Graft Rejection/immunology , T-Lymphocytes/immunology , Animals , Heart Transplantation , Kidney Transplantation , Lymphocyte Activation , Mice , Transplants
16.
J Immunol ; 197(4): 1471-6, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27357151

ABSTRACT

Pancreatic islet transplantation is a promising therapy for diabetes, but acute rejection of the islets by host effector T cells has hindered clinical application. In this study, we addressed the mechanisms of CD8(+) effector T cell migration to islet grafts because interrupting this step is key to preventing rejection. We found that effector T cell migration to revascularized islet transplants in mice is dependent on non-self Ag recognition rather than signaling via Gαi-coupled chemokine receptors. Presentation of non-self Ag by donor cells was necessary for migration, whereas Ag presentation by recipient cells was dispensable. We also observed that deficiency of SKAP1, an immune cell adaptor downstream of the TCR and important for integrin activation, prolongs allograft survival but does not reduce effector T cell migration to the graft. Therefore, effector T cell migration to transplanted islets is Ag driven, not chemokine driven, but SKAP1 does not play a critical role in this process.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte/immunology , Graft Rejection/immunology , Islets of Langerhans Transplantation/immunology , Animals , Islets of Langerhans/immunology , Mice , Models, Animal
18.
J Clin Invest ; 124(8): 3579-89, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24983319

ABSTRACT

Maturation of T cell-activating APCs directly links innate and adaptive immunity and is typically triggered by microbial infection. Transplantation of allografts, which are sterile, generates strong T cell responses; however, it is unclear how grafts induce APC maturation in the absence of microbial-derived signals. A widely accepted hypothesis is that dying cells in the graft release "danger" molecules that induce APC maturation and initiate the adaptive alloimmune response. Here, we demonstrated that danger signals associated with dying cells are not sufficient to initiate alloimmunity, but that recognition of allogeneic non-self by the innate immune system is required. In WT as well as in T cell-, B cell-, and innate lymphoid cell-deficient mice, allogeneic grafts elicited persistent differentiation of monocytes into mature DCs that expressed IL-12 and stimulated T cell proliferation and IFN-γ production. In contrast, syngeneic grafts in the same mice elicited transient and less pronounced differentiation of monocytes into DCs, which neither expressed IL-12 nor stimulated IFN-γ production. In a model in which T cell recognition is restricted to a single foreign antigen on the graft, rejection occurred only if the allogeneic non-self signal was also sensed by the host's innate immune system. These findings underscore the importance of innate recognition of allogeneic non-self by monocytes in initiating graft rejection.


Subject(s)
Graft Rejection/etiology , Monocytes/immunology , Adaptive Immunity , Allografts , Animals , B-Lymphocytes/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Dendritic Cells/immunology , Female , Graft Rejection/immunology , Heart Transplantation , Immunity, Innate , Isoantigens , Isografts , Kidney Transplantation , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction/immunology , T-Lymphocytes/immunology , Time Factors
19.
Immunol Rev ; 258(1): 145-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24517431

ABSTRACT

Vertebrates mount strong adaptive immune responses to transplanted organs (allografts), but the mechanisms by which the innate immune system initiates this response are not completely understood. In anti-microbial immunity, non-self molecules associated with pathogens but not present in the host induce the maturation of innate antigen-presenting cells (APCs) by binding to germ-line-encoded receptors. Mature APCs then initiate the adaptive immune response by presenting microbial antigen and providing costimulatory signals to T cells. How allografts activate APCs, however, is less clear, because allografts are presumably sterile. A widely accepted view is that inflammatory or 'danger' molecules released by dying graft cells at the time of transplantation trigger APC maturation and the T-cell response that follows. Alternatively, it has been proposed that the introduction of microbial products during the surgical procedure could also alert the innate immune system to the presence of the transplanted organ. Here, we review why these hypotheses fail to fully explain how the alloimmune response is initiated after transplantation and summarize evidence that recognition of allogeneic non-self by monocytes is a key event in triggering alloimmunity and graft rejection.


Subject(s)
Immunity, Innate , Organ Transplantation , Transplantation Tolerance , Allografts , Animals , Antigen-Presenting Cells/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival , Humans , Molecular Sequence Data , Organ Transplantation/adverse effects , Treatment Outcome
20.
J Clin Invest ; 123(6): 2663-71, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23676459

ABSTRACT

The migration of effector or memory T cells to the graft is a critical event in the rejection of transplanted organs. The prevailing view is that the key steps involved in T cell migration - integrin-mediated firm adhesion followed by transendothelial migration - are dependent on the activation of Gαi-coupled chemokine receptors on T cells. In contrast to this view, we demonstrated in vivo that cognate antigen was necessary for the firm adhesion and transendothelial migration of CD8+ effector T cells specific to graft antigens and that both steps occurred independent of Gαi signaling. Presentation of cognate antigen by either graft endothelial cells or bone marrow-derived APCs that extend into the capillary lumen was sufficient for T cell migration. The adhesion and transmigration of antigen-nonspecific (bystander) effector T cells, on the other hand, remained dependent on Gαi, but required the presence of antigen-specific effector T cells. These findings underscore the primary role of cognate antigen presented by either endothelial cells or bone marrow-derived APCs in the migration of T cells across endothelial barriers and have important implications for the prevention and treatment of graft rejection.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/physiology , Heart Transplantation/immunology , Kidney Transplantation/immunology , Transendothelial and Transepithelial Migration/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Adhesion , Coronary Vessels/immunology , Coronary Vessels/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Graft Rejection/immunology , Graft Rejection/prevention & control , Immunotherapy, Adoptive , Kidney/blood supply , Kidney/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Fluorescence , Myocardium/immunology , Receptors, Chemokine/physiology , Signal Transduction , Time-Lapse Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...